Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Genomics ; 105(1): 5-16, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25451739

ABSTRACT

Previously, we have shown that shortening of telomeres by telomerase inhibition sensitized cancer cells to cisplatinum, slowed their migration, increased DNA damage and impaired DNA repair. The mechanism behind these effects is not fully characterized. Its clarification could facilitate novel therapeutics development and may obviate the time consuming process of telomere shortening achieved by telomerase inhibition. Here we aimed to decipher the microRNA and proteomic profiling of cancer cells with shortened telomeres and identify the key mediators in telomere shortening-induced damage to those cells. Of 870 identified proteins, 98 were differentially expressed in shortened-telomere cells. 47 microRNAs were differentially expressed in these cells; some are implicated in growth arrest or act as oncogene repressors. The obtained data was used for a network construction, which provided us with nodal candidates that may mediate the shortened-telomere dependent features. These proteins' expression was experimentally validated, supporting their potential central role in this system.


Subject(s)
MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/metabolism , Proteome/analysis , Telomere Shortening , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Oligonucleotides/pharmacology , Proteomics , Tumor Cells, Cultured
2.
Br J Cancer ; 107(11): 1844-52, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23169337

ABSTRACT

BACKGROUND: The importance of telomerase in multiple myeloma (MM) is well established; however, its response to bortezomib has not been addressed. METHODS: The effect of bortezomib on telomerase activity and cell proliferation was evaluated in four MM cell lines and in myeloma cells obtained from eight patients. The mechanism of telomerase regulation on epigenetic, transcriptional, and post-translational levels was further assessed in two selected cell lines: ARP-1 and CAG. Clinical data were correlated with the laboratory findings. RESULTS: Bortezomib downregulated telomerase activity and decreased proliferation in all cell lines and cells obtained from patients, albeit in two different patterns of kinetics. ARP-1 cells demonstrated higher and earlier sensitivity than CAG cells due to differential phosphorylation of hTERT by PKCα. Methylation of hTERT promoter was not affected. Transcription of hTERT was similarly inhibited in both lines by decreased binding of SP-1 and not of C-Myc and NFκB. The ex vivo results confirmed the in vitro findings and suggested existence of clinical relevance. CONCLUSION: Bortezomib downregulates telomerase activity in MM cells both transcriptionally and post-translationally. MM cells, both in vitro and in patients, exhibit different sensitivity to the drug due to different post-translational response. The effect of bortezomib on telomerase activity may correlate with resistance to bortezomib in patients, suggesting its potential utility as a pre-treatment assessment.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Multiple Myeloma/drug therapy , Pyrazines/pharmacology , Telomerase/antagonists & inhibitors , Bortezomib , Cell Line, Tumor , Cell Survival/drug effects , DNA Methylation , Down-Regulation , Humans , Multiple Myeloma/enzymology , Phosphorylation , Promoter Regions, Genetic , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Telomerase/genetics
3.
Exp Oncol ; 29(2): 126-31, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17704745

ABSTRACT

UNLABELLED: Little is known about the interaction of novel anticancer drugs with other treatment modalities. THE AIM of this study was to examine the effect of combining imatinib mesylate (STI-571) with radiation or cisplatin on the survival of two human solid tumor cell lines - SKNMC cells derived from Ewing sarcoma and breast cancer MCF-7 cells. METHODS: Cell proliferation was determined using the sulphorodamine B cytotoxicity assay. Cell cycle analysis was performed with flow cytometry. Apoptosis was determined using a commercial cell death ELISA plus kit. Phosphorylated AKT, which has been suggested to be involved in radiation resistance, was detected by Western blot analysis. RESULTS: Exposure of SKNMC cells to STI-571 resulted in a dose-dependent antiproliferative effect and a decrease in phosphorylated AKT expression. There was no evidence of apoptosis. The combination of STI-571 with radiation or cisplatin had an additive antiproliferative effect in SKNMC cells (60% reduction in cell number). A similar effect was observed in human MCF-7 breast cancer cells. CONCLUSION: STI-571 improves the outcome of cisplatin or irradiation treatment in vitro. AKT pathway may play a role in the additive effect of STI-571 and irradiation.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , Sarcoma, Ewing/drug therapy , Apoptosis/drug effects , Benzamides , Blotting, Western , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Dose-Response Relationship, Drug , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Dyes/metabolism , Humans , Imatinib Mesylate , In Vitro Techniques , Inhibitory Concentration 50 , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rhodamines/metabolism , Sarcoma, Ewing/pathology , Sarcoma, Ewing/radiotherapy
4.
Br J Cancer ; 92(10): 1881-91, 2005 May 23.
Article in English | MEDLINE | ID: mdl-15870711

ABSTRACT

Imatinib mesylate (IM) is a tyrosine kinase inhibitor, which inhibits phosphorylation of downstream proteins involved in BCR-ABL signal transduction. It has proved beneficial in treating patients with chronic myeloid leukaemia (CML). In addition, IM demonstrates activity against malignant cells expressing c-kit and platelet-derived growth factor receptor (PDGF-R). The activity of IM in the blastic crisis of CML and against various myeloma cell lines suggests that this drug may also target other cellular components. In the light of the important role of telomerase in malignant transformation, we evaluated the effect of IM on telomerase activity (TA) and regulation in various malignant cell lines. Imatinib mesylate caused a dose-dependent inhibition of TA (up to 90% at a concentration of 15 microM IM) in c-kit-expressing SK-N-MC (Ewing sarcoma), SK-MEL-28 (melanoma), RPMI 8226 (myeloma), MCF-7 (breast cancer) and HSC 536/N (Fanconi anaemia) cells as well as in ba/F3 (murine pro-B cells), which do not express c-kit, BCR-ABL or PDGF-R. Imatinib mesylate did not affect the activity of other DNA polymerases. Inhibition of TA was associated with 50% inhibition of proliferation. The inhibition of proliferation was associated with a decrease in the S-phase of the cell cycle and an accumulation of cells in the G2/M phase. No apoptosis was observed. Inhibition of TA was caused mainly by post-translational modifications: dephosphorylation of AKT and, to a smaller extent, by early downregulation of hTERT (the catalytic subunit of the enzyme) transcription. Other steps of telomerase regulation were not affected by IM. This study demonstrates an additional cellular target of IM, not necessarily mediated via known tyrosine kinases, that causes inhibition of TA and cell proliferation.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Melanoma/pathology , Multiple Myeloma/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Sarcoma, Ewing/pathology , Skin Neoplasms/pathology , Telomerase/pharmacology , Animals , Benzamides , Cell Proliferation , Dose-Response Relationship, Drug , Down-Regulation , Fanconi Anemia/pathology , Humans , Imatinib Mesylate , Mice , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/pharmacology , Telomerase/biosynthesis , Tumor Cells, Cultured
5.
Endocrinology ; 144(10): 4519-26, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12960058

ABSTRACT

Experimental evidence suggested that secretion of steroid hormones from adrenocortical cells involves carrier-mediated transport: Cortisol release from, and uptake of p-[3H]aminohippurate into, bovine adrenocortical cells showed properties of the renal p-[3H]aminohippurate/anion exchanger OAT1. Other poly-specific transporters such as organic anion-transporting polypeptides (oatps) and organic cation transporters (OCTs) could also be involved in steroid hormone release. A homology-cloning procedure was established to detect these transporters in rat adrenal gland cDNA. PCR revealed the presence of OAT1, oatp1, oatp2, and oatp3. In situ hybridization localized OAT1 in the outer zona fasciculata, oatp3 in the zona glomerulosa, and oatp1 and oatp2 in the inner zona fasciculata and outer zona reticularis. An OCT2-specific probe produced signals in the zona glomerulosa and outer zona fasciculata. Pretreatment of rats with ACTH increased the expression of OAT1 mRNA that spread to all zones, and hypophysectomy strongly decreased it. A less pronounced regulation was detected for OCT2 and oatp3. Specific antibodies confirmed the localization of OAT1 in the outer zona fasciculata, supporting a possible role of OAT1 in cortisol release. The zonated distribution of transporters furthermore suggest that oatp1-3 and OCT2 may be important for the endocrine function of rat adrenocortical cells.


Subject(s)
Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/pharmacology , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Adrenal Cortex/drug effects , Animals , Hypophysectomy , In Situ Hybridization , Oocytes , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tissue Distribution/drug effects , Xenopus laevis
6.
Breast Cancer Res Treat ; 70(1): 27-37, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11767002

ABSTRACT

Recent studies from our laboratory have revealed that basic fibroblast growth factor (bFGF) selectively inhibits the proliferation of human MCF-7 breast cancer cells. It has also been shown to enhance cis-platinum-induced apoptosis, decrease levels of the anti-apoptotic gene product bcl-2, and increase levels of the cyclin-dependent protein kinase inhibitor p21/WAF1/Cip1. Transforming growth factor beta-1 (TGFbeta1), a cell growth regulator has been found to have an inhibitory effect on breast cancer cells. The aim of the present study was to evaluate the possible role of TGFbeta1 in the antiproliferative effects of bFGF in MCF-7 breast cancer cells. We found that exogenous, as well as endogenous (overexpressed) bFGF increased TGFbeta1 mRNA expression in the cells and enhanced the secretion of TGFbeta1 into culture medium. However, exogenous addition of TGFbeta1 neither led to a decrease in bcl-2 nor induced an increase in the levels of p21/WAF1/Cip1 and neutralizing antibodies to TGFbeta1, did not reverse bFGF-induced G1 arrest northe increase in p21/WAF1/Cip1 level. In contrast, antisense oligonucleotides to TGFbeta1 abrogated the antiproliferative effects and inhibited the induction of p21/WAF1/Cip1 by bFGF in MCF-7 cells. These data suggest that the anti-proliferative effects of bFGF in human MCF-7 breast cancer cells are mediated by endogenous TGFbeta1, while exogenous TGFbeta1 does not mimic all the effects of bFGF on these breast cancer cells. These findings provide an important basis for further investigations into the autocrine and paracrine processes that control the growth of breast cancer cells.


Subject(s)
Angiogenesis Inhibitors/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cyclins/metabolism , Fibroblast Growth Factor 2/metabolism , Growth Inhibitors/metabolism , Transforming Growth Factor beta/metabolism , Angiogenesis Inhibitors/pharmacology , Blotting, Northern , Blotting, Western , Breast Neoplasms/blood supply , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Gene Expression Profiling , Growth Inhibitors/genetics , Humans , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Tumor Cells, Cultured
7.
Pflugers Arch ; 438(6): 860-4, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10591075

ABSTRACT

Our study found the uptake of [14C]succinate into bovine adrenocortical cells to be sodium-dependent, inhibited by lithium, and to have an apparent K(m) of 146 mumol/l. Succinate uptake was inhibited by glutarate, fumarate, alpha-ketoglutarate, and maleate but not by 2,3-dimethylsuccinate or cis-aconitate, specific inhibitors of the basolateral Na(+)-dicarboxylate transporter of renal proximal tubule cells. Succinate uptake was highest at pH 6.0 and decreased with increasing pH. Transport of succinate was not significantly inhibited by citrate at pH 7.4 whereas at pH 6.0 inhibition of succinate uptake by citrate was small but significant. The affinity of the adrenal dicarboxylate transporter towards succinate ranges in between the low affinity of the renal luminal dicarboxylate transporter and the high affinity of the respective basolateral transporter. The pH dependency of succinate uptake and the missing inhibition by citrate at pH 7.4 differ from both the luminal and from the basolateral dicarboxylate transporters in kidney, liver, intestine, and placenta. These functional characteristics provide evidence for the existence of a Na(+)-dicarboxylate cotransporter in adrenocortical cells which may supply cholesterol metabolism with reducing substrates.


Subject(s)
Adrenal Cortex/metabolism , Carrier Proteins/metabolism , Dicarboxylic Acid Transporters , Membrane Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent , Symporters , Adrenal Cortex/cytology , Animals , Cattle , Cells, Cultured , Citric Acid/pharmacology , Hydrogen-Ion Concentration , Lithium/pharmacology , Methylation , Sodium/pharmacology , Succinates/metabolism , Succinates/pharmacology , Succinic Acid/antagonists & inhibitors , Succinic Acid/pharmacokinetics
8.
J Cancer Res Clin Oncol ; 125(10): 556-62, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10473868

ABSTRACT

Basic fibroblast growth factor (bFGF) is a classical mitogen in fibroblasts and endothelial cells. Our previous studies have demonstrated that bFGF inhibits the growth of MCF-7 human breast cancer cells. The aim of the present study was to examine the effect of bFGF on cis-diamminedichloroplatinum(cisplatin)-induced cytotoxicity in MCF-7 breast cancer cells as compared to normal endothelial cells. MCF-7/NCF cells transduced with a vector expressing the bFGF gene and overexpressing its product, and MCF-7/N2 cells transduced with the backbone vector were incubated with a combination of bFGF and cisplatin for 5 days; results were compared with those obtained with bovine aortic endothelial cells. Cell proliferation was assessed with the sulforhodamine B colorimetric cytotoxicity assay. Apoptosis was quantitatively determined by flow-cytometric analysis for DNA damage and the apoptotic death assay for DNA fragmentation, and qualitatively by electron microscopy. Reverse transcriptase/polymerase chain reaction analysis and an enzyme immunoassay were used to determine the mRNA and protein level, respectively, of the anti-apoptotic bcl-2 gene product. We found that bFGF enhanced cisplatin-induced cytotoxicity in MCF-7 breast cancer sublines. bFGF enhanced proliferation of normal endothelial cells and did not increase cisplatin-induced cytotoxicity. This effect was accompanied by down-regulation of the anti-apoptotic protooncogene bcl-2 and the enhancement of cisplatin-induced apoptosis. We suggest that the improved understanding of the role of bFGF in the differential modulation of the response of breast cancer and normal endothelial cells to chemotherapy may enable active intervention to alter the therapeutic ratio favorably in breast cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Fibroblast Growth Factor 2/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Separation , Cisplatin/therapeutic use , DNA, Neoplasm/drug effects , Drug Synergism , Female , Flow Cytometry , Humans , Microscopy, Electron , Proto-Oncogene Proteins c-bcl-2/analysis , RNA, Neoplasm/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Cell Physiol Biochem ; 9(2): 72-80, 1999.
Article in English | MEDLINE | ID: mdl-10394000

ABSTRACT

Recently we provided evidence for the involvement of a probenecid-inhibitable anion exchanger in cortisol release from primary cultures of bovine adrenocortical cells. In the present study, we further characterized this exchange transporter. Adrenocorticotropic hormone stimulated 3H-p-aminohippurate (3H-PAH) uptake into as well as cortisol release from the cells about two- and tenfold, respectively. Probenecid inhibited both 3H-PAH uptake and cortisol release by about 55 and 63%. Preincubation of the cells with 1 mM PAH trans-stimulated 3H-PAH uptake by 30%, whereas cortisol release was inhibited by 30%. 3H-PAH uptake was cis-inhibited by 1 mM glutarate or by 1 mM cortisol in the medium, while cortisol release was trans-stimulated by glutarate. PAH in the incubation medium showed saturable cis-inhibition of 3H-PAH uptake. The release of cyclic adenosine monophosphate, a substrate of the renal PAH exchanger, was also inhibited by probenecid and trans-stimulated by glutarate. In summary, the trans-stimulation and cis-inhibition experiments support the concept of an anion exchanger involved in cortisol and cyclic adenosine monophosphate release from and PAH uptake into adrenocortical cells.


Subject(s)
Adrenal Cortex/physiology , Antiporters/physiology , Cyclic AMP/metabolism , Hydrocortisone/metabolism , Probenecid/pharmacology , p-Aminohippuric Acid/pharmacokinetics , Adrenal Cortex/drug effects , Adrenocorticotropic Hormone/pharmacology , Animals , Antiporters/antagonists & inhibitors , Biological Transport/drug effects , Cattle , Cells, Cultured , Culture Media , Glutarates/pharmacology , Hydrocortisone/pharmacology , Kinetics , Tritium
10.
Neuroreport ; 9(13): 3103-7, 1998 Sep 14.
Article in English | MEDLINE | ID: mdl-9804324

ABSTRACT

Taurine is an inhibitory amino acid in the CNS. When supplied to rats it produces analgesia in some acute pain tests. Here we examined the effect of taurine supplementation on sensitivity to pain in intact rats, and whether perioperative dietary supplementation with taurine in rats would suppress autotomy, a behavior produced by peripheral neurectomy and related to neuropathic pain. Thermal pain sensitivity of intact rats consuming 1% taurine in the drinking solution for 2 weeks was not significantly different from that of control rats. Autotomy levels, determined in rats consuming taurine pre-, post- or perioperatively were significantly lower than in matching control groups. We conclude that taurine plays an important role in the autotomy model, presumably by protecting inhibitory neurons in the CNS against an excitotoxic damage triggered by injury discharge and ectopic input from the severed nerves.


Subject(s)
Amino Acids/pharmacology , Pain/drug therapy , Self Mutilation/drug therapy , Taurine/pharmacology , Administration, Oral , Animals , Disease Models, Animal , Hindlimb/innervation , Male , Pain Measurement/drug effects , Peripheral Nervous System/injuries , Peripheral Nervous System/physiopathology , Rats , Rats, Inbred Strains , Taurine/administration & dosage , Taurine/urine
11.
Biochem Pharmacol ; 56(8): 1029-34, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9776314

ABSTRACT

In this study, we evaluated the effect of several ligands active at the central-type and peripheral-type benzodiazepine receptor (BzR) (clonazepam, diazepam, PK11195 and Ro5-4864) on the growth and differentiation of B16 melanoma cells. All tested BzR ligands were able to suppress proliferation of the cells at the micromolar range and in a concentration-dependent manner. However, agents selectively active at the peripheral-type BzR (PK11195 and Ro5-4864) exhibited more potent antiproliferative activity. In addition, the BzR ligands were demonstrated to affect the cell cycle by reducing the percent of cells in the S phase and increasing the percent in the G2/M phase. BzR ligands induced cellular phenotypic alterations, which have been previously shown to be associated with melanoma cell differentiation. These alterations included: marked morphological changes, enhancement of melanogenesis, lipid accumulation and increase in the activity of gamma glutamyl transpeptidase. All BzR ligands induced a marked reduction in the concentration of UTP and most of them did the same in GTP and CTP, while ATP levels were not significantly altered. In summary, BzR ligands (clonazepam, diazepam, PK11195 and Ro5-4864) were found to exert antitumor effects in B16 melanoma cells. These findings encourage further studies of a possible therapeutic potential of BzR ligands in treatment of melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , GABA Agents/pharmacology , Melanoma, Experimental/drug therapy , Animals , Benzodiazepinones/pharmacology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Clonazepam/pharmacology , Diazepam/pharmacology , Isoquinolines/pharmacology , Melanoma, Experimental/pathology , Mice , Nucleotides/metabolism , Phenotype , Tumor Cells, Cultured , gamma-Glutamyltransferase/drug effects
12.
Isr J Med Sci ; 32(12): 1153-7, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007143

ABSTRACT

Anti-tumor effects of agents known to intervene with signal transduction pathways (ras and protein kinase c cascades) were examined in the B16 melanoma cell model. The compounds examined included: lovastatin, an inhibitor of HMG-CoA reductase, which interferes with membrane localization of p21 ras protein; H-7, a classic inhibitor of protein kinase C; and tiazofurin, a GTP depleting agent, that might affect the GTP/GDP ratio on p21ras. The three agents were found to inhibit the proliferation of B16 melanoma cells. Only tiazofurin, as expected, induced a significant decrease in GTP levels. Lovastatin and H-7 altered p21 subcellular localization. They reduced membrane expression of p21 ras, while increasing its expression in the cytosol. Following tiazofurin treatment a trend towards increased membranal p21 was observed. These results suggest that p21 is a target for the action of signal transduction inhibitors. However, the relationship between growth inhibition and altered p21 expression is not yet clear.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lovastatin/pharmacology , Proto-Oncogene Proteins p21(ras)/drug effects , Ribavirin/analogs & derivatives , Signal Transduction/drug effects , Tumor Cells, Cultured/drug effects , Cell Division/drug effects , Drug Screening Assays, Antitumor , Humans , Melanoma , Ribavirin/pharmacology , Tumor Cells, Cultured/cytology
13.
J Cancer Res Clin Oncol ; 120(12): 717-22, 1994.
Article in English | MEDLINE | ID: mdl-7798296

ABSTRACT

The effects of the differentiation-inducing agents sodium butyrate (NaOBt), dimethylsulfoxide (DMSO) and mycophenolic acid (MA), on purine nucleotide metabolism, was studied in an ovarian carcinoma cell line (GZL-8). Exposure to these agents inhibited cell proliferation, but did not affect cell viability. Three hours following exposure, NaOBt and DMSO moderately decelerated purine synthesis de novo, but MA accelerated it three-fold, this being associated with a two-fold increase in the excretion of hypoxanthine and xanthine into the incubation medium. NaOBt and DMSO did not affect the cellular nucleotide content, but MA caused a 73% decrease in GTP content and about a 50% increase in the cellular content of UTP. The following alterations in cellular enzyme activity were observed 72 h following exposure: NaOBt decreased the activity of hypoxanthine-guanine phosphoribosyltransferase and increased the activity of IMP and of AMP 5'-nucleotidases, DMSO increased the activity of IMP 5'-nucleotidase, and MA increased the activity of the two nucleotidases. The results suggest that, in the carcinoma cell line studied, the differentiation process induced by NaOBt and DMSO may be associated with a general shift in the direction of purine metabolism from anabolism to catabolism, whereas that induced by MA is associated with a specific decrease in the production of GTP.


Subject(s)
Butyrates/pharmacology , Dimethyl Sulfoxide/pharmacology , Mycophenolic Acid/pharmacology , Ovarian Neoplasms/metabolism , Purine Nucleotides/metabolism , Butyric Acid , Cell Differentiation/drug effects , Cell Division/drug effects , Female , Guanosine Triphosphate/metabolism , Humans , IMP Dehydrogenase/metabolism , Tumor Cells, Cultured
14.
J Cancer Res Clin Oncol ; 120(10): 599-604, 1994.
Article in English | MEDLINE | ID: mdl-7929531

ABSTRACT

The effect of the antibiotic agent novobiocin on the sensitivity of melanoma cells to colchicine and vinblastine was examined in drug-sensitive and drug-resistant B16 melanoma cells. A cell line COL/R was selected for colchicine resistance. The COL/R cell line (resistant to 80 ng/ml colchicine) was found to possess the multidrug-resistant (MDR) phenotype. The cells were shown to be cross-resistant to vinblastine and Adriamycin and to overexpress P glycoprotein. P glycoprotein activity was assessed by using the rhodamine 123 accumulation test. Rhodamine accumulation was markedly decreased in COL/R cells as compared to the parental B16 cells. Verapamil reversed drug resistance and increased rhodamine accumulation in COL/R cells. Novobiocin in combination with colchicine or vinblastine synergistically inhibited the proliferation of parental B16 cells. In COL/R cells, novobiocin markedly decreased colchicine resistance and increased rhodamine accumulation. These data show that novobiocin increases the sensitivity of both parental and MDR melanoma cells to microtubule-disrupting cytotoxic drugs.


Subject(s)
Colchicine/toxicity , Drug Resistance, Multiple , Novobiocin/pharmacology , Vinblastine/toxicity , Animals , Antimetabolites, Antineoplastic/metabolism , Cell Division/drug effects , Cell Line , Clone Cells , Dose-Response Relationship, Drug , Melanoma, Experimental , Mice , Phenotype , Rhodamine 123 , Rhodamines/metabolism , Tumor Cells, Cultured , Verapamil/pharmacology
15.
Cancer Invest ; 11(5): 523-9, 1993.
Article in English | MEDLINE | ID: mdl-8402221

ABSTRACT

Guanosine is shown to potentiate markedly the antiproliferative effect of cytosine-beta-D-arabinoside (ara-C) on B16 F10 mouse and SKMEL-28 human melanoma cell lines. Several metabolic consequences of the synergistic interaction between ara-C and guanosine on cell growth were determined in B16 F10 mouse melanoma cells. Treatment of the cells with guanosine for 24 hr resulted in an increase in the percentage of cells in the S phase of the cell cycle, a threefold increase in intracellular GTP concentration, and an increase in the incorporation of ara-C into acid-insoluble material and phosphorylated metabolites. These findings suggest that guanosine potentiates the growth-inhibitory effect of ara-C in B16 F10 melanoma cells by increasing the intracellular concentration of its active metabolites.


Subject(s)
Cytarabine/pharmacology , Guanosine/pharmacology , Melanoma/pathology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cytarabine/metabolism , Drug Synergism , Humans , Melanoma/metabolism , Melanoma, Experimental/pathology , Mice , Tumor Cells, Cultured
16.
Eur J Cancer ; 28(1): 22-7, 1992.
Article in English | MEDLINE | ID: mdl-1348952

ABSTRACT

A cell line (GZL-8) was established by cloning from ascitic fluid of an untreated ovarian carcinoma patient. The cells grew rapidly, accumulated lipids and showed chromosomal alterations. One of the marker chromosomes showed characteristics of a Y-like chromosome. This unusual finding was confirmed by DNA hybridisation using specific probes to the Y chromosome. The cells stained with fluorescent antibodies to desmoplakin and cytokeratins 8, 18, 19, and weakly with vimentin but not with desmin. The presence of epithelial membrane antigen, human milk fat globulin, alpha-lactalbumin, alpha-fetoprotein, placental alkaline phosphatase and oestrogen receptor-related antigen was demonstrated by indirect immunoperoxidase staining, but no CA-125 antigen could be detected. The cells showed positive reaction with antibodies to P-glycoprotein. The function of the P-glycoprotein transport system was demonstrated by the rhodamine-123 release test. The cells were initially responsive to doxorubicin, and to high concentrations of cisplatin. Growth inhibition by doxorubicin, especially at low doses was enhanced by the addition of verapamil or tamoxifen. This was shown by the soft agar clonogenic assay, by direct cell counting and by the MTT reducing test. Our results show that combination between drug and sensitivity modulators may be of potential clinical value in ovarian cancer.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Ovarian Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Antigens, Neoplasm/analysis , Carrier Proteins/metabolism , Cisplatin/pharmacology , Doxorubicin/pharmacology , Female , Humans , Karyotyping , Membrane Glycoproteins/metabolism , Tamoxifen/pharmacology , Tumor Cells, Cultured , Verapamil/pharmacology , Y Chromosome
17.
Eur J Cancer ; 26(8): 905-7, 1990.
Article in English | MEDLINE | ID: mdl-2145937

ABSTRACT

The effect of the transition metal chelator, 8-hydroxyquinoline (8-HQ), was examined on the growth and phenotype expression of B16 mouse melanoma cells. Micromolar concentrations of 8-HQ inhibited the growth of B16 cells as well as human melanoma cell lines. Removal of 8-HQ from the culture medium restored normal cell growth. Growth inhibition by 8-HQ was accompanied by phenotypic alterations that included changes in cell morphology, increased production of melanin and enhanced activities of the enzymes gamma-glutamyl transpeptidase and NADPH cytochrome c reductase. These changes might be associated with a better differentiated phenotype.


Subject(s)
Melanoma/drug therapy , Oxyquinoline/therapeutic use , Animals , Cell Division/drug effects , Cell Line , Humans , Melanoma/pathology , Melanoma, Experimental/drug therapy , Mice
18.
Eur J Cancer Clin Oncol ; 25(12): 1765-8, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2576649

ABSTRACT

The effect of sodium butyrate was examined on the growth and phenotypic expression of a cell line derived from the ascitic fluid of an untreated patient with ovarian carcinoma. The chemical inducer of differentiation, sodium butyrate, markedly enhances the activity of the membrane-bound glycoprotein enzymes, alkaline phosphatase and gamma-glutamyl transpeptidase. The alkaline phosphatase corresponds to placental Regan type. Sodium butyrate (1 mM) alone has only a small inhibitory effect on cell growth. However, it was shown to potentiate the anti-proliferative effect of Adriamycin and to render the cells sensitive to cis-platinum.


Subject(s)
Alkaline Phosphatase/metabolism , Butyrates/pharmacology , Drug Screening Assays, Antitumor , Ovarian Neoplasms/enzymology , gamma-Glutamyltransferase/metabolism , Ascitic Fluid/pathology , Butyric Acid , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Synergism , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/ultrastructure
19.
Cancer Lett ; 47(3): 193-7, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2576843

ABSTRACT

L-Histidinol, a histidine analog was recently shown to be an inducer of differentiation in the promyelocytic cell line HL-60. In the present study we show that L-histidinol inhibits the growth of B16 melanoma cells in vitro. Growth inhibition is accompanied by phenotypic alterations that include a marked increase in the activities of NADPH cytochrome c reductase and gamma glutamyl transpeptidase, lipid accumulation and cell enlargement. These phenotypic alterations are similar to those induced by other chemical inducers of differentiation in melanoma cells.


Subject(s)
Cell Division/drug effects , Histidinol/pharmacology , Imidazoles/pharmacology , Melanoma/pathology , Animals , Azo Compounds , Cell Differentiation/drug effects , Cell Line/drug effects , Histocytochemistry , Lipids/analysis , Melanoma/metabolism , Mice , NADPH-Ferrihemoprotein Reductase/analysis , Phenotype , Spectrophotometry , gamma-Glutamyltransferase/analysis
20.
Eur J Cancer Clin Oncol ; 25(5): 883-9, 1989 May.
Article in English | MEDLINE | ID: mdl-2737221

ABSTRACT

The effect of the nucleoside anti-metabolite tiazofurin (TR) was examined on the growth and phenotypic alterations of MCF-7 breast cancer and HBL-100 normal breast cell lines. TR was shown to inhibit MCF-7 cell growth. This inhibition could be reversed by exogenous addition of guanosine. The anti-proliferative effect of TR is accompanied by phenotypic alterations that include lipid accumulation and an increase in alkaline phosphatase activity. In contrast to MCF-7 cells, the HBL-100 breast milk derived cell line is relatively resistant to inhibition by TR. Alkaline phosphatase is not affected by TR and untreated cells accumulate lipid droplets, similar to TR-treated MCF-7 cells. Determination of GTP and ATP pools in both cell lines revealed that TR markedly reduces GTP content in MCF-7 cells. In HBL-100 cells, TR induces only a small decrease in GTP and does not affect ATP levels. The prototypic IMP dehydrogenase inhibitor, mycophenolic acid (MA), markedly inhibits HBL-100 cell growth, similarly to its effect on MCF-7 breast cancer cells. These findings may suggest differential metabolism of TR in MCF-7 and HBL-100 cells.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Breast Neoplasms/pathology , Milk, Human/cytology , Ribavirin/therapeutic use , Ribonucleosides/therapeutic use , Adenosine Triphosphate/analysis , Breast Neoplasms/analysis , Breast Neoplasms/drug therapy , Cell Division/drug effects , Cell Line , Female , Guanosine Triphosphate/analysis , Humans , Mycophenolic Acid/pharmacology , Ribavirin/analogs & derivatives , Tumor Cells, Cultured/analysis , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...